Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
PLoS Pathog ; 11(5): e1004884, 2015 May.
Article in English | MEDLINE | ID: mdl-26020932

ABSTRACT

The fungus Cryptococcus is a major cause of meningoencephalitis in HIV-infected as well as HIV-uninfected individuals with mortalities in developed countries of 20% and 30%, respectively. In HIV-related disease, defects in T-cell immunity are paramount, whereas there is little understanding of mechanisms of susceptibility in non-HIV related disease, especially that occurring in previously healthy adults. The present description is the first detailed immunological study of non-HIV-infected patients including those with severe central nervous system (s-CNS) disease to 1) identify mechanisms of susceptibility as well as 2) understand mechanisms underlying severe disease. Despite the expectation that, as in HIV, T-cell immunity would be deficient in such patients, cerebrospinal fluid (CSF) immunophenotyping, T-cell activation studies, soluble cytokine mapping and tissue cellular phenotyping demonstrated that patients with s-CNS disease had effective microbiological control, but displayed strong intrathecal expansion and activation of cells of both the innate and adaptive immunity including HLA-DR+ CD4+ and CD8+ cells and NK cells. These expanded CSF T cells were enriched for cryptococcal-antigen specific CD4+ cells and expressed high levels of IFN-γ as well as a lack of elevated CSF levels of typical T-cell specific Th2 cytokines -- IL-4 and IL-13. This inflammatory response was accompanied by elevated levels of CSF NFL, a marker of axonal damage, consistent with ongoing neurological damage. However, while tissue macrophage recruitment to the site of infection was intact, polarization studies of brain biopsy and autopsy specimens demonstrated an M2 macrophage polarization and poor phagocytosis of fungal cells. These studies thus expand the paradigm for cryptococcal disease susceptibility to include a prominent role for macrophage activation defects and suggest a spectrum of disease whereby severe neurological disease is characterized by immune-mediated host cell damage.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cryptococcus neoformans/immunology , Killer Cells, Natural/immunology , Meningitis, Cryptococcal/immunology , Th1 Cells/immunology , Adult , Autopsy , Brain/immunology , Brain/metabolism , Brain/pathology , CD4-Positive T-Lymphocytes/microbiology , CD8-Positive T-Lymphocytes/microbiology , Cohort Studies , Cytokines/metabolism , Female , Flow Cytometry , Humans , Immunophenotyping , Killer Cells, Natural/microbiology , Lymphocyte Activation , Male , Meningitis, Cryptococcal/cerebrospinal fluid , Meningitis, Cryptococcal/microbiology , Middle Aged , Young Adult
2.
Ann Neurol ; 78(1): 3-20, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25808056

ABSTRACT

OBJECTIVE: The management of complex patients with neuroimmunological diseases is hindered by an inability to reliably measure intrathecal inflammation. Currently implemented laboratory tests developed >40 years ago either are not dynamic or fail to capture low levels of central nervous system (CNS) inflammation. Therefore, we aimed to identify and validate biomarkers of CNS inflammation in 2 blinded, prospectively acquired cohorts of untreated patients with neuroimmunological diseases and embedded controls, with the ultimate goal of developing clinically useful tools. METHODS: Because biomarkers with maximum utility reflect immune phenotypes, we included an assessment of cell specificity in purified primary immune cells. Biomarkers were quantified by optimized electrochemiluminescent immunoassays. RESULTS: Among markers with cell-specific secretion, soluble CD27 is a validated biomarker of intrathecal T-cell activation, with an area under the receiver operating characteristic curve of 0.97. Comparing the quantities of cerebrospinal fluid (CSF) immune cells and their respective cell-specific soluble biomarkers (released by CSF cells as well as their counterparts in CNS tissue) provided invaluable information about stationary CNS immune responses, previously attainable via brain biopsy only. Unexpectedly, progressive and relapsing-remitting multiple sclerosis (MS) patients have comparable numbers of activated intrathecal T and B cells, which are preferentially embedded in CNS tissue in the former group. INTERPRETATION: The cell-specific biomarkers of intrathecal inflammation may improve diagnosis and management of neuroimmunological diseases and provide pharmacodynamic markers for future therapeutic developments in patients with intrathecal inflammation that is not captured by imaging, such as in progressive MS.


Subject(s)
Biomarkers/cerebrospinal fluid , Cerebrospinal Fluid/cytology , Multiple Sclerosis, Chronic Progressive/cerebrospinal fluid , Multiple Sclerosis, Relapsing-Remitting/cerebrospinal fluid , Adult , Aged , B-Lymphocytes/cytology , Case-Control Studies , Cerebrospinal Fluid/immunology , Cohort Studies , Female , Humans , Inflammation/cerebrospinal fluid , Interleukin-12 Subunit p40/cerebrospinal fluid , Interleukin-8/cerebrospinal fluid , Lipopolysaccharide Receptors/cerebrospinal fluid , Lymphocyte Count , Male , Middle Aged , Nervous System Diseases/cerebrospinal fluid , Prospective Studies , Receptors, Complement 3d/metabolism , T-Lymphocytes/cytology , Tumor Necrosis Factor Receptor Superfamily, Member 7/cerebrospinal fluid , Young Adult
3.
J Immunol ; 192(6): 2551-63, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24510966

ABSTRACT

We performed unbiased, comprehensive immunophenotyping of cerebrospinal fluid (CSF) and blood leukocytes in 221 subjects referred for the diagnostic work-up of neuroimmunological disorders to obtain insight about disease-specific phenotypes of intrathecal immune responses. Quantification of 14 different immune cell subsets, coupled with the assessment of their activation status, revealed physiological differences between intrathecal and systemic immunity, irrespective of final diagnosis. Our data are consistent with a model where the CNS shapes intrathecal immune responses to provide effective protection against persistent viral infections, especially by memory T cells, plasmacytoid dendritic cells, and CD56(bright) NK cells. Our data also argue that CSF immune cells do not simply reflect cells recruited from the periphery. Instead, they represent a mixture of cells that are recruited from the blood, have been activated intrathecally and leave the CNS after performing effector functions. Diagnosis-specific differences provide mechanistic insight into the disease process in the defined subtypes of multiple sclerosis (MS), neonatal onset multisystem inflammatory disease, and Aicardi-Goutières syndrome. This analysis also determined that secondary-progressive MS patients are immunologically closer to relapsing-remitting patients as compared with patients with primary-progressive MS. Because CSF immunophenotyping captures the biology of the intrathecal inflammatory processes, it has the potential to guide optimal selection of immunomodulatory therapies in individual patients and monitor their efficacy. Our study adds to the increasing number of publications that demonstrate poor correlation between systemic and intrathecal inflammatory biomarkers in patients with neuroimmunological diseases and stresses the importance of studying immune responses directly in the intrathecal compartment.


Subject(s)
Autoimmune Diseases of the Nervous System/cerebrospinal fluid , Immunophenotyping/methods , Inflammation/cerebrospinal fluid , Multiple Sclerosis, Chronic Progressive/cerebrospinal fluid , Nervous System Malformations/cerebrospinal fluid , Adolescent , Adult , Aged , Autoimmune Diseases of the Nervous System/blood , Autoimmune Diseases of the Nervous System/immunology , CD56 Antigen/immunology , CD56 Antigen/metabolism , Cell Count , Dendritic Cells/immunology , Dendritic Cells/metabolism , Flow Cytometry , Humans , Immune System/cytology , Immune System/immunology , Immune System/metabolism , Immunologic Memory/immunology , Inflammation/blood , Inflammation/immunology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Leukocytes/immunology , Leukocytes/metabolism , Middle Aged , Multiple Sclerosis, Chronic Progressive/blood , Multiple Sclerosis, Chronic Progressive/immunology , Nervous System Malformations/blood , Nervous System Malformations/immunology , Prospective Studies , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Young Adult
4.
Front Neurol ; 9: 554, 2018.
Article in English | MEDLINE | ID: mdl-30079049

ABSTRACT

Although B cell depletion is an effective therapy of multiple sclerosis (MS), the pathogenic functions of B cells in MS remain incompletely understood. We asked whether cerebrospinal fluid (CSF) B cells in MS secrete different cytokines than control-subject B cells and whether cytokine secretion affects MS phenotype. We blindly studied CSF B cells after their immortalization by Epstein-Barr Virus (EBV) in prospectively-collected MS patients and control subjects with other inflammatory-(OIND) or non-inflammatory neurological diseases (NIND) and healthy volunteers (HV). The pilot cohort (n = 80) was analyzed using intracellular cytokine staining (n = 101 B cell lines [BCL] derived from 35 out of 80 subjects). We validated differences in cytokine production in newly-generated CSF BCL (n = 207 BCL derived from subsequent 112 prospectively-recruited subjects representing validation cohort), using ELISA enhanced by objective, flow-cytometry-based B cell counting. After unblinding the pilot cohort, the immortalization efficiency was almost 5 times higher in MS patients compared to controls (p < 0.001). MS subjects' BCLs produced significantly more vascular endothelial growth factor (VEGF) compared to control BCLs. Progressive MS patients BCLs produced significantly more tumor necrosis factor (TNF)-α and lymphotoxin (LT)-α than BCL from relapsing-remitting MS (RRMS) patients. In the validation cohort, we observed lower secretion of IL-1ß in RRMS patients, compared to all other diagnostic categories. The validation cohort validated enhanced VEGF-C production by BCL from RRMS patients and higher TNF-α and LT-α secretion by BCL from progressive MS. No significant differences among diagnostic categories were observed in secretion of IL-6 or GM-CSF. However, B cell secretion of IL-1ß, TNF-α, and GM-CSF correlated significantly with the rate of accumulation of disability measured by MS disease severity scale (MS-DSS). Finally, all three cytokines with increased secretion in different stages of MS (i.e., VEGF-C, TNF-α, and LT-α) enhance lymphangiogenesis, suggesting that intrathecal B cells directly facilitate the formation of tertiary lymphoid follicles, thus compartmentalizing inflammation to the central nervous system.

5.
PLoS One ; 9(8): e105434, 2014.
Article in English | MEDLINE | ID: mdl-25148387

ABSTRACT

Multiple sclerosis (MS) is a chronic inflammatory disorder of the central nervous system (CNS). Neither the antigenic target(s) nor the cell population(s) responsible for CNS tissue destruction in MS have been fully defined. The objective of this study was to simultaneously determine the antigen (Ag)-specificity and phenotype of un-manipulated intrathecal CD4+ and CD8+ T cells of patients with relapsing-remitting and progressive MS compared to subjects with other inflammatory neurological diseases. We applied a novel Ag-recognition assay based on co-cultures of freshly obtained cerebrospinal fluid T cells and autologous dendritic cells pre-loaded with complex candidate Ag's. We observed comparably low T cell responses to complex auto-Ag's including human myelin, brain homogenate, and cell lysates of apoptotically modified oligodendroglial and neuronal cells in all cohorts and both compartments. Conversely, we detected a strong intrathecal enrichment of Epstein-Barr virus- and human herpes virus 6-specific (but not cytomegalovirus-specific) reactivities of the Th1-phenotype throughout all patients. Qualitatively, the intrathecal enrichment of herpes virus reactivities was more pronounced in MS patients. This enrichment was completely reversed by long-term treatment with the IL-2 modulating antibody daclizumab, which strongly inhibits MS disease activity. Finally, we observed a striking discrepancy between diminished intrathecal T cell proliferation and enhanced cytokine production of herpes virus-specific T cells among progressive MS patients, consistent with the phenotype of terminally differentiated cells. The data suggest that intrathecal administration of novel therapeutic agents targeting immune cells outside of the proliferation cycle may be necessary to effectively eliminate intrathecal inflammation in progressive MS.


Subject(s)
Herpesviridae Infections/complications , Herpesviridae , Multiple Sclerosis/etiology , Adult , Aged , Antibodies, Monoclonal, Humanized/pharmacology , Antigens, Viral/immunology , Autoantigens/immunology , Case-Control Studies , Daclizumab , Epitopes, T-Lymphocyte/immunology , Female , Herpesviridae/immunology , Herpesviridae Infections/immunology , Humans , Immunoglobulin G/pharmacology , Immunophenotyping , Immunosuppressive Agents/pharmacology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Male , Middle Aged , Phenotype , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Young Adult
6.
J Immunother ; 36(2): 133-51, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23377668

ABSTRACT

Nine cancer patients were treated with adoptive cell therapy using autologous anti-MAGE-A3 T-cell receptors (TCR)-engineered T cells. Five patients experienced clinical regression of their cancers including 2 on-going responders. Beginning 1-2 days postinfusion, 3 patients (#'s 5, 7, and 8) experienced mental status changes, and 2 patients (5 and 8) lapsed into comas and subsequently died. Magnetic resonance imagining analysis of patients 5 and 8 demonstrated periventricular leukomalacia, and examination of their brains at autopsy revealed necrotizing leukoencephalopathy with extensive white matter defects associated with infiltration of CD3(+)/CD8(+) T cells. Patient 7, developed Parkinson-like symptoms, which resolved over 4 weeks and fully recovered. Immunohistochemical staining of patient and normal brain samples demonstrated rare positively staining neurons with an antibody that recognizes multiple MAGE-A family members. The TCR used in this study recognized epitopes in MAGE-A3/A9/A12. Molecular assays of human brain samples using real-time quantitative-polymerase chain reaction, Nanostring quantitation, and deep-sequencing indicated that MAGE-A12 was expressed in human brain (and possibly MAGE-A1, MAGE-A8, and MAGE-A9). This previously unrecognized expression of MAGE-A12 in human brain was possibly the initiating event of a TCR-mediated inflammatory response that resulted in neuronal cell destruction and raises caution for clinical applications targeting MAGE-A family members with highly active immunotherapies.


Subject(s)
Antigens, Neoplasm , Genetic Therapy/methods , Immunotherapy, Adoptive , Melanoma/therapy , Neoplasm Proteins , Receptors, Antigen, T-Cell , Adult , Aged , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Antigens, Neoplasm/therapeutic use , Brain/metabolism , Dendritic Cells/immunology , Female , Gene Transfer Techniques , Humans , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Immunotherapy, Adoptive/mortality , Male , Melanoma/immunology , Melanoma-Specific Antigens , Middle Aged , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Neoplasm Proteins/therapeutic use , RNA, Messenger/analysis , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/therapeutic use , Vaccines, Subunit/immunology , Young Adult
7.
Nat Med ; 17(5): 604-9, 2011 May.
Article in English | MEDLINE | ID: mdl-21532597

ABSTRACT

Although previous studies have described CD25 expression and production of interleukin-2 (IL-2) by mature dendritic cells (mDCs), it remains unclear how these molecules participate in the activation of T cells. In search of the mechanisms by which daclizumab, a humanized monoclonal antibody against CD25, inhibits brain inflammation in multiple sclerosis, we observed that although the drug has limited effects on polyclonal T cell activation, it potently inhibits activation of antigen-specific T cells by mDCs. We show that mDCs (and antigen-experienced T cells) secrete IL-2 toward the mDC-T cell interface in an antigen-specific manner, and mDCs 'lend' their CD25 to primed T cells in trans to facilitate early high-affinity IL-2 signaling, which is crucial for subsequent T cell expansion and development of antigen-specific effectors. Our data reveal a previously unknown mechanism for the IL-2 receptor system in DC-mediated activation of T cells.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antigen Presentation , Dendritic Cells/immunology , Immunoglobulin G/pharmacology , Interleukin-2/immunology , T-Lymphocytes/immunology , Antibodies, Monoclonal, Humanized , Antigen Presentation/drug effects , Daclizumab , Dendritic Cells/drug effects , Humans , Immunosuppressive Agents/pharmacology , In Vitro Techniques , Interleukin-2 Receptor alpha Subunit/antagonists & inhibitors , Interleukin-2 Receptor beta Subunit/immunology , Lymphocyte Activation/drug effects , Models, Immunological , Multiple Sclerosis/drug therapy , Multiple Sclerosis/immunology , Signal Transduction , T-Lymphocytes/drug effects , Transplantation Tolerance/drug effects , Transplantation Tolerance/immunology
SELECTION OF CITATIONS
SEARCH DETAIL